Open access

Introductory Chapter: Update on Mesenchymal and Induced Pluripotent Stem Cells

Written By

Khalid Ahmed Al-Anazi

Submitted: 11 September 2019 Published: 06 February 2020

DOI: 10.5772/intechopen.90236

From the Edited Volume

Update on Mesenchymal and Induced Pluripotent Stem Cells

Edited by Khalid Ahmed Al-Anazi

Chapter metrics overview

868 Chapter Downloads

View Full Metrics

1. Introduction

Stem cells are a subset of biological cells in the human body that are capable of self-renewal, tissue repair, differentiation, and division into different cell lineages [1, 2, 3]. Based on their origin and potency, stem cells are divided into either (1) embryonic and adult (non-embryonic) stem cells or (2) unipotent, oligopotent, totipotent, multipotent, and pluripotent stem cells [1, 2, 4, 5]. Multipotent or adult stem cells include mesenchymal stem cells (MSCs), while pluripotent stem cells include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) [5].

Advertisement

2. MSCs

MSCs are heterogeneous, non-hematopoietic, adult multipotent stromal progenitor cells that are capable of self-renewal and differentiation into multiple lineages and various cell types [6, 7, 8, 9, 10, 11, 12]. They were first described in the 1960s by Alexander Friedenstein [7, 8, 10, 13]. They can be isolated from the bone marrow (BM), peripheral blood, umbilical cord blood, amniotic fluid, placenta, adipose tissue (AT), dental pulp, palatal tonsil, synovial fluid, salivary glands, as well as liver, lung, skin, and skeletal muscle tissues [6, 7, 8, 9, 10, 11, 12, 13]. The main source of MSCs is the BM although MSCs constitute only a small fraction of the total number of cells populating the BM [7, 9, 10, 11].

MSCs have certain distinguishing features: being plastic adherent and ability of differentiation into osteoblasts, adipocytes, and chondrocytes, in addition to having characteristic surface markers [6, 7, 8, 10, 11, 13, 14]. On flow cytometry, they are characteristically positive for CD105, CD73, and CD90 and negative for CD45, CD34, CD11b, CD14, CD19, CD79a, and HLA-DR [6, 7, 8, 10, 11, 13]. However, several studies have shown that MSCs obtained from BM, AT, and other sources do express CD34 surface markers [9, 15, 16, 17, 18]. MSCs can be seen in abundant numbers in the circulation under the following circumstances: stem cell mobilization with growth factors, tissue injuries, stroke, hypoxia, and inflammatory conditions [9, 19, 20, 21, 22, 23, 24]. Despite the efforts made over the last five decades including identification of nine transcriptional factors, little is known about the molecular basis underlying the stemness of MSCs, and it is still unclear whether the recently discovered genes regulate stemness or only differentiation of MSCs [12].

MSCs have immunomodulatory and immunosuppressive properties that enable them to have several therapeutic and clinical applications, which include the enhancement of engraftment as well as prevention and treatment of graft versus host disease (GVHD) in recipients of allogeneic hematopoietic stem cell transplantation (HSCT); treatment of several autoimmune disorders such as systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis, type I diabetes mellitus, and Crohn’s disease; role in regenerative medicine and tissue repair including treatment of myocardial ischemia, myocardial infarction, cardiac dysfunction, dilated cardiomyopathy, chronic non-healing wounds, critical limb ischemia, liver injury, spinal cord injuries, as well as macular degeneration, corneal reconstruction, and transplantation; neurological disorders such as multiple sclerosis and amyotrophic lateral sclerosis; bone and cartilage diseases such as osteogenesis imperfecta; and treatment of various infections and acute respiratory distress syndrome [6, 7, 11, 25, 26, 27].

MSCs are major constituents of hematopoietic stem cell niche which is a highly complex and dynamic microenvironment of the BM [28]. Leptin receptor (LepR) is a marker that enriches BM-MSCs, and LepR+ cells in the BM are a major source of bone, cartilage, and adipocytes [29]. The exosome secretome of BM-MSCs regulates stem cell maintenance and their regenerative potential, and this BM-derived secretome will be critical to the future development of therapeutic strategies for oncologic diseases and regenerative medicine [30]. Apparently, MSCs are the masters of survival and clonality as they communicate with diverse immune cells and interact with other cellular components of the BM microenvironment as well as with normal cells, leukemic stem cells, and progenitor cells [31]. The main functions of MSCs include formation of hematopoietic microenvironment, modulation of the activity of the immune system, and regulating cell trafficking [32]. When stimulated by specific signals, MSCs can be released from BM niche into circulation and can be recruited to the target tissues where they undergo in situ differentiation and contribute to tissue regeneration and homeostasis [33]. The efficacy of MSCs is linked to their immunosuppressive and anti-inflammatory properties primarily due to the release of soluble factors [34].

The putative roles of BM-MSCs during infection are detection of pathogens; activation of host immune response; elimination of pathogens; induction of proinflammatory gradients; and modulation of proinflammatory host immune response [6, 7]. Examples of the immunoregulatory properties of MSCs include inhibition of differentiation of monocytes to dendritic cells (DCs), alteration of cytokine profile of DCs, induction of tolerant phenotypes of naïve and effector T cells, inhibition of antibody production by B cells, and suppression of natural killer (NK) cell proliferation and NK-mediated cytotoxicity [35]. BM-MSCs may augment antimicrobial responses, abridge proinflammatory and damage responses, and ameliorate injury caused by the host defense to the pathogen [6, 7]. BM-MSCs appear to function as a critical fulcrum providing balance by promoting pathogen clearance during the initial inflammatory response and suppressing inflammation to preserve host integrity and facilitate tissue repair [6].

MSCs could potentially be involved at multiple levels in host defense by mobilizing immune effector cells and modulation of proinflammatory immune responses so as to minimize the tissue damage induced by inflammation [6, 36]. The immunomodulatory properties of MSCs are mediated by both: cell to cell interaction and the secreted cytokines [36, 37]. BM-MSCs may protect against infectious challenge either by direct effects on the pathogens or through indirect effects on the host [6]. On the other hand, certain types of MSCs, particularly placenta-derived MSCs and fetal membrane-derived MSCs, are highly susceptible to herpes viruses including varicella zoster virus [7, 38].

Studies have shown that several types of stem cells including BM-MSCs and neural stem cells can cross the blood brain barrier and reach tumors localized in the brain such as glioblastoma multiforme as well as ischemic areas and injured sites in the brain and engraft there. Hence, MSCs can be used as means of cellular carriers or Trojan horses to deliver cytotoxic genes or therapeutic agents for brain tumors, and they can be used to exert their therapeutic and regenerative effects in the brain [39, 40, 41, 42, 43]. In cancer, MSCs are a double-edged sword as they can exert stimulatory effects on tumor development, while they can have inhibitory effects on cancer cell growth and metastases [44]. MSCs have anticancer properties, and they can be engineered or modified to become carriers of suicide genes, employed as carriers of anti-angiogenesis factors, and utilized to target cancer stem cells [45, 46, 47]. MSCs have recently been engineered to express antiproliferative, anti-apoptotic, and antiangiogenic agents that specifically target different types of solid tumors [45].

The capacity of MSCs to proliferate and differentiate into other cells in addition to their ability to release biomolecules such as cytokines, growth factors, and microvesicles that have anti-inflammatory, immunomodulatory, anti-fibrogenic, and trophic functions make MSCs ideal candidates to function as delivery platform for cellular and gene therapies [48, 49]. Consequently, clinical trials incorporating the utilization of MSCs in the treatment of immune-related diseases have rapidly evolved after reports from preclinical studies confirming their safety and efficacy [49]. Recently, scientists have established several strategies to generate highly functional AT-derived MSCs and these include preconditioning of AT-MSCs with various stimulants and inflammatory agents; genetic manipulation of AT-MSCs; modification of culture conditions with three-dimensional aggregate formation and hypoxic culture; and proper utilization of exosome and extracellular vesicles (ECVs) that are secreted by AT-MSCs [50, 51]. Also, the main focus has recently shifted from studying differentiation of MSCs to studying their paracrine properties such as the release of ECVs that contain numerous micro-RNAs (miRNAs) including regulatory miRNAs and the production of multiple bioactive proteins and compounds that regulate MSC differentiation [52]. Hence, soluble elements derived from MSCs including ECVs have recently been proposed as a cell-free alternative for various therapies on the clinical side [51].

The combination of MSCs and tissue engineering technology can enhance the immunoregulatory properties of MSCs, and this will ultimately lead to further expansion of their utilization in regenerative medicine [53]. Tissue engineering strategies such as the use of various types of stem cells, scaffolds, medical devices, gene therapy, and nanotopography have resulted in progressing the translation of basic research towards clinical therapeutics [54, 55]. Despite the remarkable progress in MSC therapies, sufficient data on the biodistribution of MSCs, cellular and molecular structures of their target cells, and mechanisms by which MSCs reach these targets are still lacking [56]. Also, several obstacles need to be overcome before the utilization of specific types of MSCs in tissue engineering becomes a routine practice in the clinical arena [57]. Currently, human MSCs are generated through conventional static adherent cultures in the presence of fetal bovine serum or human-sourced supplements. Unfortunately, these methods are not ideal procedures to meet the future expectations of quality-assured human MSCs for clinical therapies in humans [58]. Additionally, having substantial gaps in our knowledge of the biology and therapeutic efficacy of MSCs presents major challenge to their sustainable implementation in clinical medicine [59]. Thus, optimizing the bioprocess to generate human MSCs and their products will improve efficacy and safety of stem cell therapies [58]. Also, improving the cultural environment of MSCs and selecting the appropriate scaffolds and induction factors are essential in improving the outcome of MSC-based tissue engineering [60].

Advertisement

3. iPSCs

Human iPSCs resemble human ESCs in many aspects including morphology, proliferation, differentiation potential, and pluripotency markers, but the epigenetic characteristics of human iPSCs are rather distinct [1, 2, 5, 61]. Although the utilization of iPSCs can avoid the obstacles and ethical concerns that limit the use of human ESCs, clinical application of human iPSCs still has a number of disadvantages that include chromosomal instability and tumorigenic potential, thus raising questions about the safety of their clinical utilization, and low reprogramming efficiency in addition to other concerns about their reproducibility for laboratory applications in disease modelling and drug screening [1, 3, 5, 61, 62].

In 2006, Takahashi and Yamanaka were the first scientists to generate mouse iPSCs from dermal fibroblasts through retroviral-mediated ectopic expression of the four genes: OCT4, SOX2, KLF4, and c-MYC [1, 3, 4, 63]. Since this discovery, iPSCs have been used in many research and clinical trials, including disease modelling; drug toxicity as well as drug discovery; and regenerative medicine [3, 4, 5]. Reprogramming of iPSCs should have the following crucial requirements: species such as human or mouse; cell type such as blood cell or fibroblast; factor, drug, chemical, or other protein molecules such as miRNA, DNA modifying agent, NANOG, or LIN28; vector such as retrovirus or lentivirus; and disease with specific genetic mutation [1, 4, 5, 64].

Human iPSCs have revolutionized the field of human disease modelling with an enormous potential to serve as paradigm shifting platforms for preclinical trials, personalized clinical diagnosis, and personalized drug therapy [65]. During the last 13 years, significant developments and remarkable progress have been achieved in enhancing reprogramming techniques and their efficacy, increasing safety of derived iPSCs, and developing different delivery methods [61, 62]. The ability to generate iPSCs from human somatic cells provides tremendous promises and opportunities in basic research and regenerative medicine and can provide a wide range of applications including cell-based therapies, drug screening, and disease modelling [61, 66].

The capacity of human iPSCs to retain patient-specific genomic, transcriptomic, proteomic, metabolomic, and other visualized big data information makes it possible to extend their applications beyond disease modelling into the field of personalized medicine which encompasses the adoption of novel prevention and treatment strategies based on individual variability [65]. The emergence of modern iPSC technology, with the capacity of these stem cells to undergo unlimited self-renewal and differentiation into any type of cell, has a great potential to advance translational applications including stem cell therapies and the generation of large-scale collections of cell lines for research purposes [67]. Recently, genomic editing technologies have been applied to correct the mutations in disease-specific iPSCs to create gene-corrected iPSCs that can be utilized in autologous stem cell-based therapies [64]. Nowadays, patient-specific iPSCs can be obtained by reprogramming of adult somatic cells by ectopic expression of pluripotency-associated transcription factors including OCT4, SOX2, KLF4, and c-MYC [64]. The availability of precisely generated iPSC-derived functional cells to replace or repair damaged tissues or organs will likely affect therapies of hematopoietic disorders and facilitate treatment of neurological, cardiovascular, hepatic, and retinal diseases and possibly diabetes mellitus [67]. Additionally, patient-specific iPSCs can bypass certain limitations of ESCs such as ethical concerns and immunological rejection [64]. The first clinical trial on cell-based therapy using iPSCs derived from patients to treat blindness started in Japan in September 2014 [67].

Advertisement

4. iPSC-MSCs and conclusion

MSCs derived from iPSCs (iPSC-MSCs) exhibit higher proliferation rate and less senescence than BM-MSCs, and thus the former cells are emerging as an attractive therapeutic option for obtaining a substantial population of stem cells in a sustained manner for applications in regenerative medicine [68, 69]. Several studies using human iPSC-MSCs and their exosomes in human and animal studies have shown that transplantation of these cells can produce protection of the liver against hepatic ischemia; reduction in the volume of brain infarction and preservation of neurological function after acute intracranial hemorrhage; prevention of osteonecrosis of femoral head by promotion of local angiogenesis and prevention of bone loss; facilitation of cutaneous wound healing by promotion of collagen synthesis and angiogenesis; and modulation of differentiation and function of DCs in order to support their clinical application in DC-mediated immune disorders [69, 70, 71, 72, 73]. Thus, MSCs and iPSCs may reshape the future of medical therapeutics and may eventually become curative for several chronic and intractable medical illnesses [2, 4, 5].

References

  1. 1. Menon S, Shailendra S, Renda A, Longaker M, Quarto N. An overview of direct somatic reprogramming: The ins and outs of iPSCs. International Journal of Molecular Sciences. 2016;17(1). pii: E141. DOI: 10.3390/ijms17010141
  2. 2. Al-Anazi KA. Stem cell treatments may reshape the future of medical therapeutics. Journal of Stem Cell Biology and Transplantation. 2016;1(1:1). DOI: 10.21767/2575-7725.100001
  3. 3. Diecke S, Jung SM, Lee J, Ju JH. Recent technological updates and clinical applications of induced pluripotent stem cells. The Korean Journal of Internal Medicine. 2014;29(5):547-557. DOI: 10.3904/kjim.2014.29.5.547
  4. 4. Singh VK, Kalsan M, Kumar N, Saini A, Chandra R. Induced pluripotent stem cells: Applications in regenerative medicine, disease modeling, and drug discovery. Frontiers in Cell and Development Biology. 2015;3:2. DOI: 10.3389/fcell.2015.00002
  5. 5. Al-Anazi KA. Induced pluripotent stem cells and their future therapeutic applications in hematology. Stem Cell Research and Therapy. 2015;5:258. DOI: 10.4172/2157-7633.1000258
  6. 6. Auletta JJ, Deans RJ, Bartholomew AM. Emerging roles for multipotent, bone marrow-derived stromal cells in host defense. Blood. 2012;119(8):1801-1809. DOI: 10.1182/blood-2011-10-384354
  7. 7. Al-Anazi KA, Al-Jasser AM. Mesenchymal stem cells-their antimicrobial effects and their promising future role as novel therapies of infectious complications in high risk patients. In: Demirer T, editor. Progress in Stem Cell Transplantation. IntechOpen; 2015. DOI: 10.5772/60640
  8. 8. Abdal Dayem A, Lee SB, Kim K, Lim KM, Jeon TI, Seok J, et al. Production of mesenchymal stem cells through stem cell reprogramming. International Journal of Molecular Sciences. 2019;20(8). pii: E1922. DOI: 10.3390/ijms20081922
  9. 9. Al-Anazi KA, Bakhit K, Al-Sagheir A, AlHashmi H, Abdulbaqi M, Al-Shibani Z, et al. Cure of insulin-dependent diabetes mellitus by an autologous hematopoietic stem cell transplantation performed to control multiple myeloma in a patient with chronic renal failure on regular hemodialysis. Journal of Stem Cell Biology and Transplantation. 2017;1(2:11). DOI: 10.21767/2575-7725.100011
  10. 10. Bobis S, Jarocha D, Majka M. Mesenchymal stem cells: Characteristics and clinical applications. Folia Histochemica et Cytobiologica. 2006;44(4):215-230
  11. 11. Kim N, Cho SG. Clinical applications of mesenchymal stem cells. Korean Journal of Internal Medicine. 2013;28(4):387-402. DOI: 10.3904/kjim.2013.28.4.387
  12. 12. Liu TM. Stemness of mesenchymal stem cells. Preliminary study. Journal of Stem Cell Therapy and Transplantation. 2017;1:071-073. DOI: 10.29328/journal.jsctt.1001008
  13. 13. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: An update. Cell Transplantation. 2016;25(5):829-848. DOI: 10.3727/096368915X689622
  14. 14. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317. DOI: 10.1080/14653240600855905
  15. 15. Lin CS, Ning H, Lin G, Lue TF. Is CD34 truly a negative marker for mesenchymal stromal cells? Cytotherapy. 2012;14(10):1159-1163. DOI: 10. 3109/14653249.2012.729817
  16. 16. Sidney LE, Branch MJ, Dunphy SE, Dua HS, Hopkinson A. Concise review: Evidence for CD34 as a common marker for diverse progenitors. Stem Cells. 2014;32(6):1380-1389. DOI: 10.1002/stem.1661
  17. 17. Stzepourginski I, Nigro G, Jacob JM, Dulauroy S, Sansonetti PJ, Eberl G, et al. CD34+ mesenchymal cells are a major component of the intestinal stem cells niche at homeostasis and after injury. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(4):E506-E513. DOI: 10.1073/pnas.1620059114
  18. 18. Eto H, Ishimine H, Kinoshita K, Watanabe-Susaki K, Kato H, Doi K, et al. Characterization of human adipose tissue-resident hematopoietic cell populations reveals a novel macrophage subpopulation with CD34 expression and mesenchymal multipotency. Stem Cells and Development. 2013;22(6):985-997. DOI: 10.1089/scd.2012.0442
  19. 19. Alvarez P, Carrillo E, Vélez C, Hita-Contreras F, Martínez-Amat A, Rodríguez-Serrano F, et al. Regulatory systems in bone marrow for hematopoietic stem/progenitor cells mobilization and homing. BioMed Research International. 2013;2013:312656. DOI: 10.1155/2013/312656
  20. 20. Rochefort GY, Delorme B, Lopez A, Hérault O, Bonnet P, Charbord P, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24(10):2202-2208
  21. 21. Lund TC, Tolar J, Orchard PJ. Granulocyte colony-stimulating factor mobilized CFU-F can be found in the peripheral blood but have limited expansion potential. Haematologica. 2008;93(6):908-912. DOI: 10.3324/haematol.12384
  22. 22. Gilevich IV, Fedorenko TV, Pashkova IA, Porkhanov VA, Chekhonin VP. Effects of growth factors on mobilization of mesenchymal stem cells. Bulletin of Experimental Biology and Medicine. 2017;162(5):684-686. DOI: 10.1007/s10517-017-3687-0
  23. 23. Xu L, Li G. Circulating mesenchymal stem cells and their clinical implications. Journal of Orthopaedic Translation. 2014;2(1):1-7. DOI: 10.1016/j.jot.2013.11.002
  24. 24. Koning JJ, Kooij G, de Vries HE, Nolte MA, Mebius RE. Mesenchymal stem cells are mobilized from the bone marrow during inflammation. Frontiers in Immunology. 2013;4:49. DOI: 10.3389/fimmu.2013.00049
  25. 25. Ding SSL, Subbiah SK, Khan MSA, Farhana A, Mok PL. Empowering mesenchymal stem cells for ocular degenerative disorders. International Journal of Molecular Sciences. 2019;20(7). pii: E1784. DOI: 10.3390/ijms20071784
  26. 26. Mansoor H, Ong HS, Riau AK, Stanzel TP, Mehta JS, Yam GH. Current trends and future perspective of mesenchymal stem cells and exosomes in corneal diseases. International Journal of Molecular Sciences. 2019;20(12). pii: E2853. DOI: 10.3390/ijms20122853
  27. 27. Leyendecker A Jr, Pinheiro CCG, Amano MT, Bueno DF. The use of human mesenchymal stem cells as therapeutic agents for the in vivo treatment of immune-related diseases: A systematic review. Frontiers in Immunology. 2018;9:2056. DOI: 10.3389/fimmu.2018.02056
  28. 28. Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, et al. PDGFRα and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. Journal of Experimental Medicine. 2013;210(7):1351-1367. DOI: 10.1084/jem.20122252
  29. 29. Zhou BO, Yue R, Murphy MM, Peyer JG, Morrison SJ. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014;15(2):154-168. DOI: 10.1016/j.stem.2014.06.008
  30. 30. Eltoukhy HS, Sinha G, Moore CA, Gergues M, Rameshwar P. Secretome within the bone marrow microenvironment: A basis for mesenchymal stem cell treatment and role in cancer dormancy. Biochimie. 2018;155:92-103. DOI: 10.1016/j.biochi.2018.05.018
  31. 31. Pleyer L, Valent P, Greil R. Mesenchymal stem and progenitor cells in normal and dysplastic hematopoiesis-masters of survival and clonality? International Journal of Molecular Sciences. 2017;17(7). pii: E1009. DOI: 10.3390/ijms17071009
  32. 32. Shi C. Recent progress toward understanding the physiological function of bone marrow mesenchymal stem cells. Immunology. 2012;136(2):133-138. DOI: 10.1111/j.1365-2567.2012.03567.x
  33. 33. Liu ZJ, Zhuge Y, Velazquez OC. Trafficking and differentiation of mesenchymal stem cells. Journal of Cellular Biochemistry. 2009;106(6):984-991. DOI: 10.1002/jcb.22091
  34. 34. De Luca L, Trino S, Laurenzana I, Lamorte D, Caivano A, Del Vecchio L, et al. Mesenchymal stem cell derived extracellular vesicles: A role in hematopoietic transplantation? International Journal of Molecular Sciences. 2017;18(5). pii: E1022. DOI: 10.3390/ijms18051022
  35. 35. Thanunchai M, Hongeng S, Thitithanyanont A. Mesenchymal stromal cells and viral infection. Stem Cells International. 2015;2015:860950. DOI: 10.1155/2015/860950
  36. 36. Castro-Manrreza ME, Montesinos JJ. Immunoregulation by mesenchymal stem cells: Biological aspects and clinical applications. Journal of Immunology Research. 2015;2015:394917. DOI: 10.1155/2015/394917
  37. 37. Kyurkchiev D, Bochev I, Ivanova-Todorova E, Mourdjeva M, Oreshkova T, Belemezova K, et al. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World Journal of Stem Cells. 2014;6(5):552-570. DOI: 10.4252/wjsc.v6.i5.552
  38. 38. Avanzi S, Leoni V, Rotola A, Alviano F, Solimando L, Lanzoni G, et al. Susceptibility of human placenta derived mesenchymal stromal/stem cells to human herpesviruses infection. PLoS ONE. 2013;8(8):e71412. DOI: 10.1371/journal.pone.0071412
  39. 39. Abdi Z, Eskandary H, Nematollahi-Mahani SN. Effects of two types of human cells on outgrowth of human glioma in rats. Turkish Neurosurgery. 2018;28(1):19-28. DOI: 10.5137/1019-5149.JTN.18697-16.1
  40. 40. Dong HJ, Li G, Meng HP, Shang CZ, Luo Y, Wen G, et al. How can mesenchymal stem cells penetrate the blood brain barrier? Turkish Neurosurgery. 2018;28(6):1013-1014. DOI: 10.5137/1019-5149.JTN.22639-18.1
  41. 41. Conaty P, Sherman LS, Naaldijk Y, Ulrich H, Stolzing A, Rameshwar P. Methods of mesenchymal stem cell homing to the blood-brain barrier. Methods in Molecular Biology. 2018;1842:81-91. DOI: 10.1007/978-1-4939-8697-2_6
  42. 42. Liu L, Eckert MA, Riazifar H, Kang DK, Agalliu D, Zhao W. From blood to the brain: Can systemically transplanted mesenchymal stem cells cross the blood-brain barrier? Stem Cells International. 2013;2013:435093. DOI: 10.1155/2013/435093
  43. 43. Christodoulou I, Goulielmaki M, Devetzi M, Panagiotidis M, Koliakos G, Zoumpourlis V. Mesenchymal stem cells in preclinical cancer cytotherapy: A systematic review. Stem Cell Research & Therapy. 2018;9(1):336. DOI: 10.1186/s13287-018-1078-8
  44. 44. Lee HY, Hong IS. Double-edged sword of mesenchymal stem cells: Cancer-promoting versus therapeutic potential. Cancer Science. 2017;108(10):1939-1946. DOI: 10.1111/cas.13334
  45. 45. Nowakowski A, Drela K, Rozycka J, Janowski M, Lukomska B. Engineered mesenchymal stem cells as an anti-cancer trojan horse. Stem Cells and Development. 2016;25(20):1513-1531. DOI: 10.1089/scd.2016.0120
  46. 46. Zhang L, Su XS, Ye JS, Wang YY, Guan Z, Yin YF. Bone marrow mesenchymal stem cells suppress metastatic tumor development in mouse by modulating immune system. Stem Cell Research & Therapy. 2015;6:45. DOI: 10.1186/s13287-015-0039-8
  47. 47. Fakiruddin KS, Ghazalli N, Lim MN, Zakaria Z, Abdullah S. Mesenchymal stem cell expressing TRAIL as targeted therapy against sensitised tumor. International Journal of Molecular Sciences. 2018;19(8). pii: E2188. DOI: 10. 3390/ijms19082188
  48. 48. D’souza N, Rossignoli F, Golinelli G, Grisendi G, Spano C, Candini O, et al. Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies. BMC Medicine. 2015;13:186. DOI: 10.1186/s12916-015-0426-0
  49. 49. Wang M, Yuan Q , Xie L. Mesenchymal stem cell-based immunomodulation: Properties and clinical application. Stem Cells International. 2018;2018:3057624. DOI: 10.1155/2018/3057624
  50. 50. Seo Y, Shin TH, Kim HS. Current strategies to enhance adipose stem cell function: An update. International Journal of Molecular Sciences. 2019;20(15). pii: E3827. DOI: 10.3390/ijms20153827
  51. 51. Najar M, Bouhtit F, Melki R, Afif H, Hamal A, Fahmi H, et al. Mesenchymal stromal cell-based therapy: New perspectives and challenges. Journal of Clinical Medicine. 2019;8(5). pii: E626. DOI: 10.3390/jcm8050626
  52. 52. Lukomska B, Stanaszek L, Zuba-Surma E, Legosz P, Sarzynska S, Drela K. Challenges and controversies in human mesenchymal stem cell therapy. Stem Cells International. 2019;2019:9628536. DOI: 10.1155/2019/9628536
  53. 53. Li H, Shen S, Fu H, Wang Z, Li X, Sui X, et al. Immunomodulatory functions of mesenchymal stem cells in tissue engineering. Stem Cells International. 2019;2019:9671206. DOI: 10.1155/2019/9671206
  54. 54. Salmasi S, Kalaskar DM, Yoon WW, Blunn GW, Seifalian AM. Role of nanotopography in the development of tissue engineered 3D organs and tissues using mesenchymal stem cells. World Journal of Stem Cells. 2015;7(2):266-280. DOI: 10.4252/wjsc.v7.i2.266
  55. 55. Fang L, Shi XF, Sun HY, Li YX, Xiao FJ, Wang H, et al. Effects of exosomes derived from miR-486 gene modified umbilical cord mesenchymal stem cells on biological characteristics of rat cardiomyocytes. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2018;26(5):1531-1537. DOI: 10.7534/j.issn.1009-2137.2018.05.045
  56. 56. Leibacher J, Henschler R. Biodistribution, migration and homing of systemically applied mesenchymal stem/stromal cells. Stem Cell Research & Therapy. 2016;7:7. DOI: 10.1186/s13287-015-0271-2
  57. 57. Nancarrow-Lei R, Mafi P, Mafi R, Khan W. A systemic review of adult mesenchymal stem cell sources and their multilineage differentiation potential relevant to musculoskeletal tissue repair and regeneration. Current Stem Cell Research & Therapy. 2017;12(8):601-610. DOI: 10.2174/1574888X12666170608124303
  58. 58. Panchalingam KM, Jung S, Rosenberg L, Behie LA. Bioprocessing strategies for the large-scale production of human mesenchymal stem cells: A review. Stem Cell Research & Therapy. 2015;6:225. DOI: 10.1186/s13287-015-0228-5
  59. 59. Schäfer R. Advanced cell therapeutics are changing the clinical landscape: Will mesenchymal stromal cells be a part of it? BMC Medicine. 2019;17(1):53. DOI: 10.1186/s12916-019-1289-6
  60. 60. Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal stem cells for regenerative medicine. Cell. 2019;8(8). pii: E866. DOI: 10.3390/cells8080886
  61. 61. Brouwer M, Zhou H, Nadif Kasri N. Choices for induction of pluripotency: Recent developments in human induced pluripotent stem cell reprogramming strategies. Stem Cell Reviews and Reports. 2016;12(1):54-72. DOI: 10.1007/s12015-015-9622-8
  62. 62. Hong SG, Dunbar CE, Winkler T. Assessing the risks of genotoxicity in the therapeutic development of induced pluripotent stem cells. Molecular Therapy. 2013;21(2):272-281. DOI: 10.1038/mt.2012.255
  63. 63. Wu Y-Y, Chiu F-L, Yeh C-S, Kuo H-C. Opportunities and challenges for the use of induced pluripotent stem cells in modelling neurodegenerative disease. Open Biology. 2019;9:180177. DOI: 10.1098/rsob.180177
  64. 64. Wattanapanitch M. Recent updates on induced pluripotent stem cells in hematological disorders. Stem Cells International. 2019;2019:5171032. DOI: 10.1155/2019/5171032
  65. 65. Matsa E, Ahrens JH, Wu JC. Human induced pluripotent stem cells as a platform for personalized and precision cardiovascular medicine. Physiological Reviews. 2016;96(3):1093-1126. DOI: 10.1152/physrev.00036.2015
  66. 66. Singh VK, Kumar N, Kalsan M, Saini A, Chandra R. Mechanism of induction: Induced pluripotent stem cells (iPSCs). Journal of Stem Cells. 2015;10(1):43-62
  67. 67. Zhou H, Martinez H, Sun B, Li A, Zimmer M, Katsanis N, et al. Rapid and efficient generation of transgene-free iPSC from a small volume of cryopreserved blood. Stem Cell Reviews and Reports. 2015;11(4):652-665. DOI: 10.1007/s12015-015-9586-8
  68. 68. Sabapathy V, Kumar S. hiPSC-derived iMSCs: NextGen MSCs as an advanced therapeutically active cell resource for regenerative medicine. Journal of Cellular and Molecular Medicine. 2016;20(8):1571-1588. DOI: 10.1111/jcmm.12839
  69. 69. Gao WX, Sun YQ , Shi J, Li CL, Fang SB, Wang D, et al. Effects of mesenchymal stem cells from human induced pluripotent stem cells on differentiation, maturation, and function of dendritic cells. Stem Cell Research & Therapy. 2017;8(1):48. DOI: 10.1186/s13287-017-0499-0
  70. 70. Du Y, Li D, Han C, Wu H, Xu L, Zhang M, et al. Exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect liver against hepatic ischemia/reperfusion injury via activating sphingosine kinase and sphingosine-1-phosphate signaling pathway. Cellular Physiology and Biochemistry. 2017;43(2):611-625. DOI: 10.1159/000480533
  71. 71. Chen KH, Lin KC, Wallace CG, Li YC, Shao PL, Chiang JY, et al. Human induced pluripotent stem cell-derived mesenchymal stem cell therapy effectively reduced brain infarct volume and preserved neurological function in rat after acute intracranial hemorrhage. American Journal of Translational Research. 2019;11(9):6232-6248
  72. 72. Liu X, Li Q , Niu X, Hu B, Chen S, Song W, et al. Exosomes secreted from human-induced pluripotent stem cell-derived mesenchymal stem cells prevent osteonecrosis of the femoral head by promoting angiogenesis. International Journal of Biological Sciences. 2017;13(2):232-244. DOI: 10.7150/ijbs.16951
  73. 73. Zhang J, Guan J, Niu X, Hu G, Guo S, Li Q , et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. Journal of Translational Medicine. 2015;13:49. DOI: 10.1186/s12967-015-0417-0

Written By

Khalid Ahmed Al-Anazi

Submitted: 11 September 2019 Published: 06 February 2020